1
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Filannino FM, Ruggiero M, Panaro MA, Lofrumento DD, Trotta T, Benameur T, Cianciulli A, Calvello R, Zoila F, Porro C. Irisin Attenuates Neuroinflammation Targeting the NLRP3 Inflammasome. Molecules 2024; 29:5623. [PMID: 39683782 DOI: 10.3390/molecules29235623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Neuroinflammation is defined as an immune response involving various cell types, particularly microglia, which monitor the neuroimmune axis. Microglia activate in two distinct ways: M1, which is pro-inflammatory and capable of inducing phagocytosis and releasing pro-inflammatory factors, and M2, which has anti-inflammatory properties. Inflammasomes are large protein complexes that form in response to internal danger signals, activating caspase-1 and leading to the release of pro-inflammatory cytokines such as interleukin 1β. Irisin, a peptide primarily released by muscles during exercise, was examined for its effects on BV2 microglial cells in vitro. Even at low concentrations, irisin was observed to influence the NLRP3 inflammasome, showing potential as a neuroprotective and anti-inflammatory agent after stimulation with lipopolysaccharides (LPSs). Irisin helped maintain microglia in their typical physiological state and reduced their migratory capacity. Irisin also increased Arg-1 protein expression, a marker of M2 polarization, while downregulating NLRP3, Pycard, caspase-1, IL-1β, and CD14. The results of this study indicate that irisin may serve as a crucial mediator of neuroprotection, thus representing an innovative tool for the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Melania Ruggiero
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, I-73100 Lecce, Italy
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| | - Tarek Benameur
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Rosa Calvello
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Federico Zoila
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| |
Collapse
|
3
|
Tastan B, Heneka MT. The impact of neuroinflammation on neuronal integrity. Immunol Rev 2024; 327:8-32. [PMID: 39470038 DOI: 10.1111/imr.13419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Neuroinflammation, characterized by a complex interplay among innate and adaptive immune responses within the central nervous system (CNS), is crucial in responding to infections, injuries, and disease pathologies. However, the dysregulation of the neuroinflammatory response could significantly affect neurons in terms of function and structure, leading to profound health implications. Although tremendous progress has been made in understanding the relationship between neuroinflammatory processes and alterations in neuronal integrity, the specific implications concerning both structure and function have not been extensively covered, with the exception of perspectives on glial activation and neurodegeneration. Thus, this review aims to provide a comprehensive overview of the multifaceted interactions among neurons and key inflammatory players, exploring mechanisms through which inflammation influences neuronal functionality and structural integrity in the CNS. Further, it will discuss how these inflammatory mechanisms lead to impairment in neuronal functions and architecture and highlight the consequences caused by dysregulated neuronal functions, such as cognitive dysfunction and mood disorders. By integrating insights from recent research findings, this review will enhance our understanding of the neuroinflammatory landscape and set the stage for future interventions that could transform current approaches to preserve neuronal integrity and function in CNS-related inflammatory conditions.
Collapse
Affiliation(s)
- Bora Tastan
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, Massachusetts, USA
| |
Collapse
|
4
|
Han Q, Li W, Chen P, Wang L, Bao X, Huang R, Liu G, Chen X. Microglial NLRP3 inflammasome-mediated neuroinflammation and therapeutic strategies in depression. Neural Regen Res 2024; 19:1890-1898. [PMID: 38227513 DOI: 10.4103/1673-5374.390964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/22/2023] [Indexed: 01/17/2024] Open
Abstract
Previous studies have demonstrated a bidirectional relationship between inflammation and depression. Activation of the nucleotide-binding oligomerization domain, leucine-rich repeat, and NLR family pyrin domain-containing 3 (NLRP3) inflammasomes is closely related to the pathogenesis of various neurological diseases. In patients with major depressive disorder, NLRP3 inflammasome levels are significantly elevated. Understanding the role that NLRP3 inflammasome-mediated neuroinflammation plays in the pathogenesis of depression may be beneficial for future therapeutic strategies. In this review, we aimed to elucidate the mechanisms that lead to the activation of the NLRP3 inflammasome in depression as well as to provide insight into therapeutic strategies that target the NLRP3 inflammasome. Moreover, we outlined various therapeutic strategies that target the NLRP3 inflammasome, including NLRP3 inflammatory pathway inhibitors, natural compounds, and other therapeutic compounds that have been shown to be effective in treating depression. Additionally, we summarized the application of NLRP3 inflammasome inhibitors in clinical trials related to depression. Currently, there is a scarcity of clinical trials dedicated to investigating the applications of NLRP3 inflammasome inhibitors in depression treatment. The modulation of NLRP3 inflammasomes in microglia holds promise for the management of depression. Further investigations are necessary to ascertain the efficacy and safety of these therapeutic approaches as potential novel antidepressant treatments.
Collapse
Affiliation(s)
- Qiuqin Han
- Department of Scientific Research, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Wenhui Li
- Department of Scientific Research, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Peiqing Chen
- Department of Scientific Research, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Lijuan Wang
- Department of Scientific Research, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xiwen Bao
- Department of Scientific Research, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Renyan Huang
- Department of Traditional Chinese Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guobin Liu
- Department of Traditional Chinese Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaorong Chen
- Department of Physiology, Laboratory of Neurodegenerative Diseases, Changzhi Medical College, Changzhi, Shanxi Province, China
| |
Collapse
|
5
|
Islam MR, Rabbi MA, Hossain T, Sultana S, Uddin S. Mechanistic Approach to Immunity and Immunotherapy of Alzheimer's Disease: A Review. ACS Chem Neurosci 2024. [PMID: 39173186 DOI: 10.1021/acschemneuro.4c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative condition characterized by progressive cognitive decline and memory loss, affecting millions of people worldwide. Traditional treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, offer limited symptomatic relief without addressing the underlying disease mechanisms. These limitations have driven the development of more potent and effective therapies. Recent advances in immunotherapy present promising avenues for AD treatment. Immunotherapy strategies, including both active and passive approaches, harness the immune system to target and mitigate AD-related pathology. Active immunotherapy stimulates the patient's immune response to produce antibodies against AD-specific antigens, while passive immunotherapy involves administering preformed antibodies or immune cells that specifically target amyloid-β (Aβ) or tau proteins. Monoclonal antibodies, such as aducanumab and lecanemab, have shown potential in reducing Aβ plaques and slowing cognitive decline in clinical trials, despite challenges related to adverse immune responses and the need for precise targeting. This comprehensive review explores the role of the immune system in AD, evaluates the current successes and limitations of immunotherapeutic approaches, and discusses future directions for enhancing the treatment efficacy.
Collapse
Affiliation(s)
- Md Rubiath Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Md Afser Rabbi
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tanbir Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Sadia Sultana
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Shihab Uddin
- Department of Bioengineering, King Fahad University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Bio Systems and Machines, King Fahad University of Petroleum & Minerals, Dhahran-31261, Saudi Arabia
| |
Collapse
|
6
|
Ravichandran KA, Heneka MT. Inflammasomes in neurological disorders - mechanisms and therapeutic potential. Nat Rev Neurol 2024; 20:67-83. [PMID: 38195712 DOI: 10.1038/s41582-023-00915-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 01/11/2024]
Abstract
Inflammasomes are molecular scaffolds that are activated by damage-associated and pathogen-associated molecular patterns and form a key element of innate immune responses. Consequently, the involvement of inflammasomes in several diseases that are characterized by inflammatory processes, such as multiple sclerosis, is widely appreciated. However, many other neurological conditions, including Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, stroke, epilepsy, traumatic brain injury, sepsis-associated encephalopathy and neurological sequelae of COVID-19, all involve persistent inflammation in the brain, and increasing evidence suggests that inflammasome activation contributes to disease progression in these conditions. Understanding the biology and mechanisms of inflammasome activation is, therefore, crucial for the development of inflammasome-targeted therapies for neurological conditions. In this Review, we present the current evidence for and understanding of inflammasome activation in neurological diseases and discuss current and potential interventional strategies that target inflammasome activation to mitigate its pathological consequences.
Collapse
Affiliation(s)
- Kishore Aravind Ravichandran
- Department of Neuroinflammation, Institute of innate immunity, University of Bonn Medical Center Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Esch-sur-Alzette, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, MA, USA.
| |
Collapse
|
7
|
Zhang L, Yao Q, Hu J, Qiu B, Xiao Y, Zhang Q, Zeng Y, Zheng S, Zhang Y, Wan Y, Zheng X, Zeng Q. Hotspots and trends of microglia in Alzheimer's disease: a bibliometric analysis during 2000-2022. Eur J Med Res 2024; 29:75. [PMID: 38268044 PMCID: PMC10807212 DOI: 10.1186/s40001-023-01602-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/17/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Alzheimer's disease is one common type of dementia. Numerous studies have suggested a correlation between Alzheimer's disease and inflammation. Microglia mainly participate in the inflammatory response in the brain. Currently, ample evidence has shown that microglia are closely related to the occurrence and development of Alzheimer's disease. OBJECTIVE We opted for bibliometric analysis to comprehensively summarize the advancements in the study of microglia in Alzheimer's disease, aiming to provide researchers with current trends and future research directions. METHODS All articles and reviews pertaining to microglia in Alzheimer's disease from 2000 to 2022 were downloaded through Web of Science Core Collection. The results were subjected to bibliometric analysis using VOSviewer 1.6.18 and CiteSpace 6.1 R2. RESULTS Overall, 7449 publications were included. The number of publications was increasing yearly. The United States has published the most publications. Harvard Medical School has published the most papers of all institutions. Journal of Alzheimer's Disease and Journal of Neuroscience were the journals with the most studies and the most commonly cited, respectively. Mt Heneka is the author with the highest productivity and co-citation. After analysis, the most common keywords are neuroinflammation, amyloid-beta, inflammation, neurodegeneration. Gut microbiota, extracellular vesicle, dysfunction and meta-analysis are the hotspots of research at the present stage and are likely to continue. CONCLUSION NLRP3 inflammasome, TREM2, gut microbiota, mitochondrial dysfunction, exosomes are research hotspots. The relationship between microglia-mediated neuroinflammation and Alzheimer's disease have been the focus of current research and the development trend of future research.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Qiuru Yao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Jinjing Hu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Baizhi Qiu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Yupeng Xiao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Qi Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Yuting Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuqi Zheng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Youao Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yantong Wan
- College of Anesthesiology, Southern Medical University, Guangzhou, China.
| | - Xiaoyan Zheng
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China.
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Hu Z, Luo Y, Zhu J, Jiang D, Luo Z, Wu L, Li J, Peng S, Hu J. Role of the P2 × 7 receptor in neurodegenerative diseases and its pharmacological properties. Cell Biosci 2023; 13:225. [PMID: 38093352 PMCID: PMC10720200 DOI: 10.1186/s13578-023-01161-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 11/02/2023] [Indexed: 12/17/2023] Open
Abstract
Neurodegenerative diseases seriously affect patients' physical and mental health, reduce their quality of life, and impose a heavy burden on society. However, their treatment remains challenging. Therefore, exploring factors potentially related to the pathogenesis of neurodegenerative diseases and improving their diagnosis and treatment are urgently needed. Recent studies have shown that P2 × 7R plays a crucial role in regulating neurodegenerative diseases caused by neuroinflammation. P2 × 7R is an adenosine 5'-triphosphate ligand-gated cation channel receptor present in most tissues of the human body. An increase in P2 × 7R levels can affect the progression of neurodegenerative diseases, and the inhibition of P2 × 7R can alleviate neurodegenerative diseases. In this review, we comprehensively describe the biological characteristics (structure, distribution, and function) of this gene, focusing on its potential association with neurodegenerative diseases, and we discuss the pharmacological effects of drugs (P2 × 7R inhibitors) used to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Ziyan Hu
- Department of the second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Yifan Luo
- Department of the second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Jinxi Zhu
- Department of the second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Danling Jiang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhenzhong Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Lidong Wu
- Department of Emergency medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jin Li
- Department of Emergency medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Jialing Hu
- Department of Emergency medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
9
|
Khilazheva ED, Mosiagina AI, Panina YA, Belozor OS, Komleva YK. Impact of NLRP3 Depletion on Aging-Related Metaflammation, Cognitive Function, and Social Behavior in Mice. Int J Mol Sci 2023; 24:16580. [PMID: 38068904 PMCID: PMC10705877 DOI: 10.3390/ijms242316580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Immunosenescence and chronic inflammation associated with old age accompany brain aging and the loss of complex behaviors. Neuroinflammation in the hippocampus plays a pivotal role in the development of cognitive impairment and anxiety. However, the underlying mechanisms have not been fully explained. In this study, we aimed to investigate the disruption of insulin signaling and the mechanisms underlying metabolic inflammation ("metaflammation") in the brains of wild-type (WT) and NLRP3 knockout (KO) mice of different ages. We found a significant upregulation of the NLRP3 inflammasome in the hippocampus during aging, leading to an increase in the expression of phosphorylated metaflammation proteinases and inflammatory markers, along with an increase in the number of senescent cells. Additionally, metaflammation causes anxiety and impairs social preference behavior in aged mice. On the other hand, deletion of NLRP3 improves some behavioral and biochemical characteristics associated with aging, such as signal memory, neuroinflammation, and metabolic inflammation, but not anxious behavior. These results are associated with reduced IL-18 signaling and the PKR/IKKβ/IRS1 pathway as well as the SASP phenotype. In NLRP3 gene deletion conditions, PKR is down-regulated. Therefore, it is likely that slowing aging through various NLRP3 inhibition mechanisms will lessen the corresponding cognitive decline with aging. Thus, the genetic knockout of the NLRP3 inflammasome can be seen as a new therapeutic strategy for slowing down central nervous system (CNS) aging.
Collapse
Affiliation(s)
- Elena D. Khilazheva
- Research Institute of Molecular Medicine and Pathobiochemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia (A.I.M.)
- Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Angelina I. Mosiagina
- Research Institute of Molecular Medicine and Pathobiochemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia (A.I.M.)
| | - Yulia A. Panina
- Research Institute of Molecular Medicine and Pathobiochemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia (A.I.M.)
- Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Olga S. Belozor
- Research Institute of Molecular Medicine and Pathobiochemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia (A.I.M.)
- Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Yulia K. Komleva
- Research Institute of Molecular Medicine and Pathobiochemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia (A.I.M.)
- Department of Biochemistry, Medical, Pharmaceutical and Toxicological Chemistry, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| |
Collapse
|
10
|
Yang H, Song R, Xie Y, Qian Q, Wu Z, Han S, Li X. Apple Polyphenol Extract Ameliorates Atherosclerosis and Associated Cognitive Impairment through Alleviating Neuroinflammation by Weakening TLR4 Signaling and NLRP3 Inflammasome in High-Fat/Cholesterol Diet-Fed LDLR -/- Male Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15506-15521. [PMID: 37824601 DOI: 10.1021/acs.jafc.3c01966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Although studies have supported the beneficial effects of the ingredients of apple polyphenol extract (APE), a polyphenol mixture being extracted from whole fresh apples, on neurodegenerative diseases, the role of APE in atherosclerosis-related cognitive impairment remains unclear. To clarify the role of APE in regulating cognitive dysfunction in mice with atherosclerosis and the underlying mechanisms, high-fat/cholesterol diet-fed male LDLR-/- mice were gavaged with 125 or 500 mg/(kg·bw·d) APE solution or sterile double-distilled water for consecutive 8 weeks, and age-matched C57BL/6 male mice were employed as normal control. APE intervention increased the serum concentration of high-density apolipoprotein cholesterol, improved atherosclerosis, and ameliorated cognitive function of mice by inhibiting the phosphorylation of tau protein, supporting with significantly reduced platform latency and obviously increased swimming distance in the target quadrant according to the Morris water maze test. APE intervention alleviated neuroinflammation by attenuating the activation of microglia and astrocytes and inhibiting TLR4 signaling with reduced protein expression of NF-κB, MyD88, TRIF, and IKKβ. Meanwhile, APE intervention inactivated NLRP3 inflammasome with downregulated protein expression of caspase-1, IL-18, and IL-1β. Additionally, APE intervention improved the damaged brain barrier structure by upregulating the protein expression of ZO-1 and occludin. Therefore, our research supplemented new data, supporting the potential of APE as an effective dietary bioactive ingredient to improve atherosclerosis and associated cognitive impairment.
Collapse
Affiliation(s)
- Hao Yang
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Ruijuan Song
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Yisha Xie
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Qingfan Qian
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Zhengli Wu
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Shufen Han
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Xinli Li
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P. R. China
| |
Collapse
|
11
|
Shen H, Song H, Wang S, Su D, Sun Q. NEAT1 enhances MPP + -induced pyroptosis in a cell model of Parkinson's disease via targeting miR-5047/YAF2 signaling. Immun Inflamm Dis 2023; 11:e817. [PMID: 37382256 PMCID: PMC10288836 DOI: 10.1002/iid3.817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/11/2023] [Accepted: 03/07/2023] [Indexed: 06/30/2023] Open
Abstract
PURPOSE Parkinson's disease (PD) is the second most frequent neurodegenerative disease. The aim of our study is to explore the role and the regulatory mechanism of long noncoding RNA (lncRNA) NEAT1 in MPP+ -induced pyroptosis in a cell model of PD. MATERIALS AND METHODS MPP+ -treated SH-SY5Y cells were used as an in vitro model of dopaminergic neurons for PD. Expression levels of miR-5047 and YAF2 mRNA were determined through qRT-PCR. TUNEL staining was carried out to analyze neuronal apoptosis. Luciferase activity assay was accomplished to analyze the combination of miR-5047 with NEAT1 or YAF2 3'-UTR region. Besides, concentrations of IL-1β and IL-18 in supernatant samples were analyzed by using ELISA assay. Expression level of proteins were examined through Western blot. RESULTS NEAT1 and YAF2 expression were increased, while miR-5047 expression was declined in the SH-SY5Y cells treated with MPP+ . NEAT1 was a positively regulator to SH-SY5Y cells pyroptosis induced by MPP+ . In addition, YAF2 was a downstream target of miR-5047. NEAT1 promoted YAF2 expression via inhibiting miR-5047. Importantly, the promotion of NEAT1 to SH-SY5Y cells pyroptosis induced by MPP+ was rescued by miR-5047 mimic transfection or YAF2 downregulation. CONCLUSION In conclusion, NEAT1 was increased in MPP+ -induced SH-SY5Y cells, and it promoted MPP+ -induced pyroptosis through facilitating YAF2 expression by sponging miR-5047.
Collapse
Affiliation(s)
- Hong Shen
- Department of EncephalopathySecond People's HospitalSuzhou CityJiangsu ProvinceChina
| | - Hui Song
- Department of Neurology, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| | - Songlin Wang
- Department of Neurology, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| | - Daojing Su
- Department of Orthopaedic Rehabilitation, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| | - Qiang Sun
- Department of Neurology, TaiHe HospitalHubei University of MedicineShiyan CityHubei ProvinceChina
| |
Collapse
|
12
|
Friedland RP, Haribabu B. Neurodegenerative diseases: from gut-brain axis to brain microbiome. Front Aging Neurosci 2023; 15:1171955. [PMID: 37273657 PMCID: PMC10237314 DOI: 10.3389/fnagi.2023.1171955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Affiliation(s)
- Robert P. Friedland
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
13
|
Cyr B, de Rivero Vaccari JP. Sex Differences in the Inflammatory Profile in the Brain of Young and Aged Mice. Cells 2023; 12:1372. [PMID: 37408205 DOI: 10.3390/cells12101372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 07/07/2023] Open
Abstract
Neurodegenerative diseases are a leading cause of death worldwide with no cures identified. Thus, there is a critical need for preventative measures and treatments as the number of patients is expected to increase. Many neurodegenerative diseases have sex-biased prevalence, indicating a need to examine sex differences when investigating prevention and treatment strategies. Inflammation is a key contributor to many neurodegenerative diseases and is a promising target for prevention since inflammation increases with age, which is known as inflammaging. Here, we analyzed the protein expression levels of cytokines, chemokines, and inflammasome signaling proteins in the cortex of young and aged male and female mice. Our results show an increase in caspase-1, interleukin (IL)-1β, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and ASC specks in females compared to males. Additionally, there was an increase in IL-1α, VEGF-A, CCL3, CXCL1, CCL4, CCL17, and CCL22 in aging females and an increase in IL-8, IL-17a, IL-7, LT-α, and CCL22 in aging males. IL-12/IL-23p40, CCL13, and IL-10 were increased in females compared to males but not with age. These results indicate that there are sex differences in cortical inflammaging and provide potential targets to attenuate inflammation to prevent the development of neurodegenerative disease.
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
14
|
Inflammasome activation in traumatic brain injury and Alzheimer's disease. Transl Res 2023; 254:1-12. [PMID: 36070840 DOI: 10.1016/j.trsl.2022.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/21/2022]
Abstract
Traumatic brain injury (TBI) and Alzheimer's disease (AD) represent 2 of the largest sources of death and disability in the United States. Recent studies have identified TBI as a potential risk factor for AD development, and numerous reports have shown that TBI is linked with AD associated protein expression during the acute phase of injury, suggesting an interplay between the 2 pathologies. The inflammasome is a multi-protein complex that plays a role in both TBI and AD pathologies, and is characterized by inflammatory cytokine release and pyroptotic cell death. Products of inflammasome signaling pathways activate microglia and astrocytes, which attempt to resolve pathological inflammation caused by inflammatory cytokine release and phagocytosis of cellular debris. Although the initial phase of the inflammatory response in the nervous system is beneficial, recent evidence has emerged that the heightened inflammatory response after trauma is self-perpetuating and results in additional damage in the central nervous system. Inflammasome-induced cytokines and inflammasome signaling proteins released from activated microglia interact with AD associated proteins and exacerbate AD pathological progression and cellular damage. Additionally, multiple genetic mutations associated with AD development alter microglia inflammatory activity, increasing and perpetuating inflammatory cell damage. In this review, we discuss the pathologies of TBI and AD and how they are impacted by and potentially interact through inflammasome activity and signaling proteins. We discuss current clinical trials that target the inflammasome to reduce heightened inflammation associated with these disorders.
Collapse
|
15
|
Forloni G. Alpha Synuclein: Neurodegeneration and Inflammation. Int J Mol Sci 2023; 24:ijms24065914. [PMID: 36982988 PMCID: PMC10059798 DOI: 10.3390/ijms24065914] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Alpha-Synuclein (α-Syn) is one of the most important molecules involved in the pathogenesis of Parkinson's disease and related disorders, synucleinopathies, but also in several other neurodegenerative disorders with a more elusive role. This review analyzes the activities of α-Syn, in different conformational states, monomeric, oligomeric and fibrils, in relation to neuronal dysfunction. The neuronal damage induced by α-Syn in various conformers will be analyzed in relation to its capacity to spread the intracellular aggregation seeds with a prion-like mechanism. In view of the prominent role of inflammation in virtually all neurodegenerative disorders, the activity of α-Syn will also be illustrated considering its influence on glial reactivity. We and others have described the interaction between general inflammation and cerebral dysfunctional activity of α-Syn. Differences in microglia and astrocyte activation have also been observed when in vivo the presence of α-Syn oligomers has been combined with a lasting peripheral inflammatory effect. The reactivity of microglia was amplified, while astrocytes were damaged by the double stimulus, opening new perspectives for the control of inflammation in synucleinopathies. Starting from our studies in experimental models, we extended the perspective to find useful pointers to orient future research and potential therapeutic strategies in neurodegenerative disorders.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy
| |
Collapse
|
16
|
Weaver DF. Alzheimer's disease as an innate autoimmune disease (AD 2): A new molecular paradigm. Alzheimers Dement 2023; 19:1086-1098. [PMID: 36165334 DOI: 10.1002/alz.12789] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/13/2022] [Accepted: 08/09/2022] [Indexed: 11/07/2022]
Abstract
A new model of Alzheimer's disease (AD) is presented: Alzheimer's disease as an autoimmune disease (AD2). In response to pathogen-/damage-associated molecular pattern-stimulating events (e.g., infection, trauma, ischemia, pollution), amyloid beta (Aβ) is released as an early responder cytokine triggering an innate immunity cascade in which Aβ exhibits immunomodulatory/antimicrobial duality. However, Aβ's antimicrobial properties result in a misdirected attack upon "self" neurons, arising from the electrophysiological similarities between neurons and bacteria in terms of transmembrane potential gradients and anionic charges on outer membrane macromolecules. The subsequent breakdown products of necrotic neurons elicit further release of Aβ leading to a chronic, self-perpetuating cycle. In AD2, amino acid (trp, arg) metabolism is a central control player in modulating AD autoimmunity. AD2 includes Aβ as an important molecular player, but rejects the "amyloid hypothesis," recognizing Aβ as a physiologically oligomerizing cytokine and part of a larger immunopathic conceptualization of AD.
Collapse
Affiliation(s)
- Donald F Weaver
- Krembil Brain Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Li M, Qian M, Jiang Q, Tan B, Yin Y, Han X. Evidence of Flavonoids on Disease Prevention. Antioxidants (Basel) 2023; 12:antiox12020527. [PMID: 36830086 PMCID: PMC9952065 DOI: 10.3390/antiox12020527] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
A growing body of evidence highlights the properties of flavonoids in natural foods for disease prevention. Due to their antioxidative, anti-inflammatory, and anti-carcinogenic activities, flavonoids have been revealed to benefit skeletal muscle, liver, pancreas, adipocytes, and neural cells. In this review, we introduced the basic classification, natural sources, and biochemical properties of flavonoids, then summarize the experimental results and underlying molecular mechanisms concerning the effects of flavonoid consumption on obesity, cancers, and neurogenerative diseases that greatly threaten public health. Especially, the dosage and duration of flavonoids intervening in these diseases are discussed, which might guide healthy dietary habits for people of different physical status.
Collapse
Affiliation(s)
- Meng Li
- Hainan Institute, Zhejiang University, Sanya 572000, China
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengqi Qian
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qian Jiang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xinyan Han
- Hainan Institute, Zhejiang University, Sanya 572000, China
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Correspondence: ; Tel.: +86-0571-88982446
| |
Collapse
|
18
|
Filippini A, Salvi V, Dattilo V, Magri C, Castrezzati S, Veerhuis R, Bosisio D, Gennarelli M, Russo I. LRRK2 Kinase Inhibition Attenuates Astrocytic Activation in Response to Amyloid β 1-42 Fibrils. Biomolecules 2023; 13:biom13020307. [PMID: 36830676 PMCID: PMC9953366 DOI: 10.3390/biom13020307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Intracerebral accumulation of amyloid-β in the extracellular plaques of Alzheimer's disease (AD) brains represents the main cause of reactive astrogliosis and neuroinflammatory response. Of relevance, leucine-rich repeat kinase 2 (LRRK2), a kinase linked to genetic and sporadic Parkinson's disease (PD), has been identified as a positive mediator of neuroinflammation upon different inflammatory stimuli, however its pathogenicity in AD remains mainly unexplored. In this study, by using pharmacological inhibition of LRRK2 and murine primary astrocytes, we explored whether LRRK2 regulates astrocytic activation in response to amyloid-β1-42 (Aβ1-42). Our results showed that murine primary astrocytes become reactive and recruit serine 935 phosphorylated LRRK2 upon Aβ1-42 fibril exposure. Moreover, we found that pharmacological inhibition of LRRK2, with two different kinase inhibitors, can attenuate Aβ1-42-mediated inflammation and favor the clearance of Aβ1-42 fibrils in astrocytes. Overall, our findings report that LRRK2 kinase activity modulates astrocytic reactivity and functions in the presence of Aβ1-42 deposits and indicate that PD-linked LRRK2 might contribute to AD-related neuroinflammation and pathogenesis.
Collapse
Affiliation(s)
- Alice Filippini
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Valentina Salvi
- Oncology and Experimental Immunology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Vincenzo Dattilo
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Chiara Magri
- Biology and Genetics Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Stefania Castrezzati
- Human Anatomy Unit, Department of Biomedical Sciences and Biotechnologies, University of Brescia, 25123 Brescia, Italy
| | - Robert Veerhuis
- Amsterdam UMC, Psychiatry, Amsterdam Public Health Research Institute and Neuroscience Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam UMC, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Daniela Bosisio
- Oncology and Experimental Immunology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Massimo Gennarelli
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Biology and Genetics Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Russo
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Biology and Genetics Unit, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Correspondence: ; Tel.: +39-030-371-7461; Fax: +39-030-370-1157
| |
Collapse
|
19
|
Roseborough AD, Zhu Y, Zhao L, Laviolette SR, Pasternak SH, Whitehead SN. Fibrinogen primes the microglial NLRP3 inflammasome and propagates pro-inflammatory signaling via extracellular vesicles: Implications for blood-brain barrier dysfunction. Neurobiol Dis 2023; 177:106001. [PMID: 36646389 DOI: 10.1016/j.nbd.2023.106001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
The brain's response to acute injury is characterized by increased permeability of the blood-brain barrier (BBB) and pro-inflammatory microglia signaling, both of which have been linked to poor cognitive outcomes and neurological disease. The damaged BBB has increased leakiness, allowing serum proteins like fibrinogen into the brain, which interacts with local cells in a deleterious manner. At the same time, in response to injury, microglia demonstrate increased NLRP3 inflammasome activity and heightened release of pro-inflammatory cytokines. The relationship between increased fibrinogen uptake and microglial inflammasome signaling in the injured brain has not been well described. In this work, we investigate fibrinogen mediated NLRP3 inflammasome priming of BV-2 cells and primary adult microglia and propose a role for extracellular vesicles (EVs) as propagators of this interaction. Following exposure to fibrinogen microglia significantly upregulate transcription of IL-1β, IL-6, NLRP3 and other pro-inflammatory cytokines which was sustained by repeated fibrinogen exposure. Inhibition of fibrinogen mediated NLRP3 signaling was achieved at the transcriptional and assembly level using cannabidiol (CBD) and the NLRP3 inhibitor MCC950, respectively. EVs released following NLRP3 priming carry IL-1β, IL-18 mRNA and fibrinogen, propagate inflammatory signaling and can be detected in the circulation following BBB disruption in a preclinical stroke model. In conclusion, the interplay between fibrinogen extravasation, microglial NLRP3 signaling, and EV release can perpetuate chronic pro-inflammatory signaling and represents a novel method of inflammatory propagation.
Collapse
Affiliation(s)
- A D Roseborough
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Y Zhu
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - L Zhao
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - S R Laviolette
- Addictions Research Group, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Psychiatry, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - S H Pasternak
- Department of Clinical Neurological Sciences, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada; Robarts Research Institute, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - S N Whitehead
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
20
|
Kattan D, Barsa C, Mekhijian S, Shakkour Z, Jammoul M, Doumit M, Zabala MCP, Darwiche N, Eid AH, Mechref Y, Wang KK, de Rivero Vaccari JP, Munoz Pareja JC, Kobeissy F. Inflammasomes as biomarkers and therapeutic targets in traumatic brain injury and related-neurodegenerative diseases: A comprehensive overview. Neurosci Biobehav Rev 2023; 144:104969. [PMID: 36423707 PMCID: PMC9805531 DOI: 10.1016/j.neubiorev.2022.104969] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022]
Abstract
Given the ambiguity surrounding traumatic brain injury (TBI) pathophysiology and the lack of any Food and Drug Administration (FDA)-approved neurotherapeutic drugs, there is an increasing need to better understand the mechanisms of TBI. Recently, the roles of inflammasomes have been highlighted as both potential therapeutic targets and diagnostic markers in different neurodegenerative disorders. Indeed, inflammasome activation plays a pivotal function in the central nervous system (CNS) response to many neurological conditions, as well as to several neurodegenerative disorders, specifically, TBI. This comprehensive review summarizes and critically discusses the mechanisms that govern the activation and assembly of inflammasome complexes and the major methods used to study inflammasome activation in TBI and its implication for other neurodegenerative disorders. Also, we will review how inflammasome activation is critical in CNS homeostasis and pathogenesis, and how it can impact chronic TBI sequalae and increase the risk of developing neurodegenerative diseases. Additionally, we discuss the recent updates on inflammasome-related biomarkers and the potential to utilize inflammasomes as putative therapeutic targets that hold the potential to better diagnose and treat subjects with TBI.
Collapse
Affiliation(s)
- Dania Kattan
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Chloe Barsa
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Sarin Mekhijian
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon; Program for Interdisciplinary Neuroscience, Department of Child Health, School of Medicine, University of Missouri, USA
| | - Maya Jammoul
- Department of Anatomy, Cell Biology, and Physiology, American University of Beirut, Beirut, Lebanon
| | - Mark Doumit
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Maria Camila Pareja Zabala
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K Wang
- Morehouse School of Medicine, Department of Neurobiology, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Jennifer C Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon; Morehouse School of Medicine, Department of Neurobiology, Atlanta, GA, USA.
| |
Collapse
|
21
|
Galea E, Graeber MB. Neuroinflammation: The Abused Concept. ASN Neuro 2023; 15:17590914231197523. [PMID: 37647500 PMCID: PMC10469255 DOI: 10.1177/17590914231197523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Scientific progress requires the relentless correction of errors and refinement of hypotheses. Clarity of terminology is essential for clarity of thought and proper experimental interrogation of nature. Therefore, the application of the same scientific term to different and even conflicting phenomena and concepts is not useful and must be corrected. Such abuse of terminology has happened and is still increasing in the case of "neuroinflammation," a term that until the 1990s meant classical inflammation affecting the central nervous system (CNS) and thereon was progressively used to mostly denote microglia activation. The resulting confusion is very wasteful and detrimental not only for scientists but also for patients, given the numerous failed clinical trials in acute and chronic CNS diseases over the last decade with "anti-inflammatory" drugs. Despite this failure, reassessments of the "neuroinflammation" concept are rare, especially considering the number of articles still using the term. This undesirable situation motivates this article. We review the origins and evolution of the term "neuroinflammation," discuss the unique tissue defense and repair strategies in the CNS, define CNS immunity, and emphasize the notion of gliopathies to help readdress, if not bury, the term "neuroinflammation" as it stands in the way of scientific progress.
Collapse
Affiliation(s)
- Elena Galea
- Departament de Bioquímica, Unitat de Bioquímica, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
- ICREA, Barcelona, Spain
| | - Manuel B. Graeber
- Faculty of Medicine and Health, Brain and Mind Centre, The University of Sydney, Camperdown, Australia
| |
Collapse
|
22
|
Lo J, Liu CC, Li YS, Lee PY, Liu PL, Wu PC, Lin TC, Chen CS, Chiu CC, Lai YH, Chang YC, Wu HE, Chen YR, Huang YK, Huang SP, Wang SC, Li CY. Punicalagin Attenuates LPS-Induced Inflammation and ROS Production in Microglia by Inhibiting the MAPK/NF-κB Signaling Pathway and NLRP3 Inflammasome Activation. J Inflamm Res 2022; 15:5347-5359. [PMID: 36131784 PMCID: PMC9484772 DOI: 10.2147/jir.s372773] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Neurodegenerative diseases are associated with neuroinflammation along with activation of microglia and oxidative stress, but currently lack effective treatments. Punicalagin is a natural bio-sourced product that exhibits anti-inflammatory effects on several chronic diseases; however, the anti-inflammatory and anti-oxidative effects on microglia have not been well examined. This study aimed to investigate the effects of punicalagin on LPS-induced inflammatory responses, NLRP3 inflammasome activation, and the production of ROS using murine microglia BV2 cells. Methods BV2 cells were pre-treated with punicalagin following LPS treatment to induce inflammation. The secretion of NO and PGE2 was analyzed by Griess reagent and ELISA respectively, while the expressions of iNOS, COX-2, STAT3, ERK, JNK, and p38 were analyzed using Western blotting, the production of IL-6 was measured by ELISA, and the activity of NF-κB was detected using promoter reporter assay. To examine whether punicalagin affects NLRP3 inflammasome activation, BV2 cells were stimulated with LPS and then treated with ATP or nigericin. The secretion of IL-1β was measured by ELISA. The expressions of NLRP3 inflammasome-related proteins and phospho IκBα/IκBα were analyzed using Western blotting. The production of intracellular and mitochondrial ROS was analyzed by flow cytometry. Results Our results showed that punicalagin attenuated inflammation with reduction of pro-inflammatory mediators and cytokines including iNOS, COX-2, IL-1β, and reduction of IL-6 led to inhibition of STAT3 phosphorylation by LPS-induced BV2 cells. Punicalagin also suppressed the ERK, JNK, and p38 phosphorylation, attenuated NF-κB activity, inhibited the activation of the NLRP3 inflammasome, and reduced the production of intracellular and mitochondrial ROS by LPS-induced BV2 cells. Conclusion Our results demonstrated that punicalagin attenuated LPS-induced inflammation through suppressing the expression of iNOS and COX-2, inhibited the activation of MAPK/NF-κB signaling pathway and NLRP3 inflammasome, and reduced the production of ROS in microglia, suggesting that punicalagin might have the potential in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Jung Lo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Ching-Chih Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, Chi Mei Medical Center, Tainan, 71004, Taiwan
| | - Yueh-Shan Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Po-Yen Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Pei-Chang Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Tzu-Chieh Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yu-Hung Lai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yo-Chen Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Hsin-En Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yuan-Ru Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yu-Kai Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Shu-Pin Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Ph.D. Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan.,Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, 91201, Taiwan
| |
Collapse
|
23
|
Forloni G, La Vitola P, Balducci C. Oligomeropathies, inflammation and prion protein binding. Front Neurosci 2022; 16:822420. [PMID: 36081661 PMCID: PMC9445368 DOI: 10.3389/fnins.2022.822420] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The central role of oligomers, small soluble aggregates of misfolded proteins, in the pathogenesis of neurodegenerative disorders is recognized in numerous experimental conditions and is compatible with clinical evidence. To underline this concept, some years ago we coined the term oligomeropathies to define the common mechanism of action of protein misfolding diseases like Alzheimer, Parkinson or prion diseases. Using simple experimental conditions, with direct application of synthetic β amyloid or α-synuclein oligomers intraventricularly at micromolar concentrations, we could detect differences and similarities in the biological consequences. The two oligomer species affected cognitive behavior, neuronal dysfunction and cerebral inflammatory reactions with distinct mechanisms. In these experimental conditions the proposed mediatory role of cellular prion protein in oligomer activities was not confirmed. Together with oligomers, inflammation at different levels can be important early in neurodegenerative disorders; both β amyloid and α-synuclein oligomers induce inflammation and its control strongly affects neuronal dysfunction. This review summarizes our studies with β-amyloid or α-synuclein oligomers, also considering the potential curative role of doxycycline, a well-known antibiotic with anti-amyloidogenic and anti-inflammatory activities. These actions are analyzed in terms of the therapeutic prospects.
Collapse
|
24
|
Wang N, Chu F, Zhang L, Fei C, Yu C, Xue S, Wang Y, Fang L, Peng D, Duan X, Chen W. Taohong siwu decoction attenuates AIM2 and NLRC4 inflammasomes by ameliorates deoxyribonucleic acid damage after ischemic stroke. Front Pharmacol 2022; 13:954867. [PMID: 36034843 PMCID: PMC9411787 DOI: 10.3389/fphar.2022.954867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
Taohong siwu decoction (THSWD) has been shown to have a therapeutic effect on ischemic strokes (IS). However, it is not clear to us whether THSWD reduces deoxyribonucleic acid (DNA) damage after stroke and reduces the inflammatory response caused by the damage. Therefore, we constructed an IS model (I/R) in rats and performed oxygen-glucose deprivation/reoxygenation (OGD/R) on BV2 cells. Then ELISA, immunofluorescence staining, immunohistochemistry staining, and RT-qPCR were performed to detect the expressions of absent in melanoma 2 (AIM2), NLRC4, and Caspase-1 inflammasomes and other inflammatory factors. Experimental stroke causes DNA damage, and we found that the aforementioned inflammasomes as well as inflammatory factors were significantly inhibited after treatment with THSWD by comparing the model group with the model administration group. In addition, we examined the expression of AIM2, NLRC4, and Caspase-1 in BV2 cells of OGD/R and found that the expression of the aforementioned inflammasomes was significantly decreased in OGD/R by administration of THSWD-containing serum. Our data suggest that THSWD can reduced DNA damage after stroke as well as the inflammatory response caused by the damage.
Collapse
Affiliation(s)
- Ni Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Key Laboratory of Xin’an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Furui Chu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Key Laboratory of Xin’an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
| | - Lijuan Zhang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Changyi Fei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Chao Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Sujun Xue
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yongzhong Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Ling Fang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Xianchun Duan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Key Laboratory of Xin’an Medicine (Anhui University of Chinese Medicine), Ministry of Education, Hefei, China
- *Correspondence: Xianchun Duan, ; Weidong Chen,
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- *Correspondence: Xianchun Duan, ; Weidong Chen,
| |
Collapse
|
25
|
Zhou X, Venigalla M, Raju R, Münch G. Pharmacological considerations for treating neuroinflammation with curcumin in Alzheimer's disease. J Neural Transm (Vienna) 2022; 129:755-771. [PMID: 35294663 DOI: 10.1007/s00702-022-02480-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 12/14/2022]
Abstract
Prof. Dr. Peter Riederer, the former Head of the Neurochemistry Department of the Psychiatry and Psychotherapy Clinic at the University of Würzburg (Germany), has been one of the pioneers of research into oxidative stress in Parkinson's and Alzheimer's disease (AD). This review will outline how his scientific contribution to the field has opened a new direction for AD treatment beyond "plaques and tangles". In the 1990s, Prof. Riederer was one of the first scientists who proposed oxidative stress and neuroinflammation as one of the major contributors to Alzheimer's disease, despite the overwhelming support for the "amyloid-only" hypothesis at the time, which postulated that the sole and only cause of AD is β-amyloid. His group also highlighted the role of advanced glycation end products, sugar and dicarbonyl-derived protein modifications, which crosslink proteins into insoluble aggregates and potent pro-inflammatory activators of microglia. For the treatment of chronic neuroinflammation, he and his group suggested that the most appropriate drug class would be cytokine-suppressive anti-inflammatory drugs (CSAIDs) which have a broader anti-inflammatory action range than conventional non-steroidal anti-inflammatory drugs. One of the most potent CSAIDs is curcumin, but it suffers from a variety of pharmacokinetic disadvantages including low bioavailability, which might have tainted many human clinical trials. Although a variety of oral formulations with increased bioavailability have been developed, curcumin's absorption after oral delivery is too low to reach therapeutic concentrations in the micromolar range in the systemic circulation and the brain. This review will conclude with evidence that rectally applied suppositories might be the best alternatives to oral medications, as this route will be able to evade first-pass metabolism in the liver and achieve high concentrations of curcumin in plasma and tissues, including the brain.
Collapse
Affiliation(s)
- Xian Zhou
- NICM Health Research Institute, Western Sydney University, 158-160 Hawkesbury Rd, Westmead, NSW, 2145, Australia
| | - Madhuri Venigalla
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Ritesh Raju
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Gerald Münch
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia.
| |
Collapse
|